Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Commun Signal ; 19(1): 70, 2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34215258

RESUMO

BACKGROUND: The requirement of promyelocytic leukaemia protein (PML) in interferon (IFN)-induced cell apoptosis is well-established. However, the exact mechanisms by which the multiple isoforms of PML protein participate in this process remain not well-understood. We previously demonstrated that PML isoform II (PML-II) positively regulates induced gene expression during a type I IFN response and evaluate here how PML-II contributes to IFNα-induced cell death. METHODS: HeLa cells were transiently depleted of PML-II by siRNA treatment and the response of these cells to treatment with IFNα assessed by molecular assays of mRNA and proteins associated with IFN and apoptosis responses. RESULTS: In HeLa cells, death during IFNα stimulation was reduced by prior PML-II depletion. PML-II removal also considerably decreased the induced expression of pro-apoptotic ISGs such as ISG54 (IFIT2), and substantially impaired or prevented expression of PUMA and TRAIL, proteins that are associated with the intrinsic and extrinsic apoptotic pathways respectively. Thirdly, PML-II depletion enhanced ERK and AKT pro-survival signaling activation suggesting that PML-II normally suppresses signaling via these pathways, and that lack of PML-II hence led to greater than normal activation of AKT signaling upon IFNα stimulation and consequently increased resistance to IFNα-induced apoptosis. CONCLUSIONS: The positive contribution of PML-II to the expression of various IFNα-induced pro-apoptotic proteins and its inhibition of pro-survival signaling together provide a mechanistic explanation for reduced apoptosis under conditions of PML deficiency and may account for at least part of the role of PML as a tumor suppressor gene. Video Abstract.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Interferon-alfa/genética , Neoplasias/genética , Proteína da Leucemia Promielocítica/genética , Proteínas de Ligação a RNA/genética , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases/genética , Neoplasias/patologia , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/genética
2.
Sci Rep ; 10(1): 8903, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483257

RESUMO

Bacterial neonatal meningitis results in high mortality and morbidity rates for those affected. Although improvements in diagnosis and treatment have led to a decline in mortality rates, morbidity rates have remained relatively unchanged. Bacterial resistance to antibiotics in this clinical setting further underlines the need for developing other technologies, such as phage therapy. We exploited an in vitro phage therapy model for studying bacterial neonatal meningitis based on Escherichia coli (E. coli) EV36, bacteriophage (phage) K1F and human cerebral microvascular endothelial cells (hCMECs). We show that phage K1F is phagocytosed and degraded by constitutive- and PAMP-dependent LC3-assisted phagocytosis and does not induce expression of inflammatory cytokines TNFα, IL-6, IL-8 or IFNß. Additionally, we observed that phage K1F temporarily decreases the barrier resistance of hCMEC cultures, a property that influences the barrier permeability, which could facilitate the transition of immune cells across the endothelial vessel in vivo. Collectively, we demonstrate that phage K1F can infect intracellular E. coli EV36 within hCMECs without themselves eliciting an inflammatory or defensive response. This study illustrates the potential of phage therapy targeting infections such as bacterial neonatal meningitis and is an important step for the continued development of phage therapy targeting antibiotic-resistant bacterial infections generally.


Assuntos
Bacteriófagos/fisiologia , Encéfalo/citologia , Endotélio Vascular/citologia , Escherichia coli/virologia , Encéfalo/metabolismo , Encéfalo/microbiologia , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/microbiologia , Endotélio Vascular/metabolismo , Endotélio Vascular/microbiologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/terapia , Adesões Focais/metabolismo , Humanos , Meningites Bacterianas/metabolismo , Meningites Bacterianas/terapia , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Terapia por Fagos , Fagocitose
3.
Commun Biol ; 3(1): 124, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32170151

RESUMO

Viral genomes have high gene densities and complex transcription strategies rendering transcriptome analysis through short-read RNA-seq approaches problematic. Adenovirus transcription and splicing is especially complex. We used long-read direct RNA sequencing to study adenovirus transcription and splicing during infection. This revealed a previously unappreciated complexity of alternative splicing and potential for secondary initiating codon usage. Moreover, we find that most viral transcripts tend to shorten polyadenylation lengths as infection progresses. Development of an open reading frame centric bioinformatics analysis pipeline provided a deeper quantitative and qualitative understanding of adenovirus's genetic potential. Across the viral genome adenovirus makes multiple distinctly spliced transcripts that code for the same protein. Over 11,000 different splicing patterns were recorded across the viral genome, most occurring at low levels. This low-level use of alternative splicing patterns potentially enables the virus to maximise its coding potential over evolutionary timescales.


Assuntos
Adenovírus Humanos/genética , Processamento Alternativo/genética , Evolução Molecular , Transcriptoma , Sequência de Bases , Linhagem Celular , Uso do Códon , Biologia Computacional/métodos , Éxons , Fibroblastos/virologia , Perfilação da Expressão Gênica , Genoma Viral , Humanos , Poliadenilação , Regiões Promotoras Genéticas , RNA Viral/genética , RNA-Seq
4.
J Gen Virol ; 97(8): 1955-1967, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27217299

RESUMO

Promyelocytic leukemia (PML) proteins have been implicated in antiviral responses but PML and associated proteins are also suggested to support virus replication. One isoform, PML-II, is required for efficient transcription of interferon and interferon-responsive genes. We therefore investigated the PML-II contribution to human adenovirus 5 (Ad5) infection, using shRNA-mediated knockdown. HelaΔII cells showed a 2-3-fold elevation in Ad5 yield, reflecting an increase in late gene expression. This increase was found to be due in part to the reduced innate immune response consequent upon PML-II depletion. However, the effect was minor because the viral E4 Orf3 protein targets and inactivates this PML-II function. The major benefit to Ad5 in HelaΔII cells was exerted via an increase in HSP70; depletion of HSP70 completely reversed this replicative advantage. Increased Ad5 late gene expression was not due either to the previously described inhibition of inflammatory responses by HSP70 or to effects of HSP70 on major late promoter or L4 promoter activity, but might be linked to an observed increase in E1B 55K, as this protein is known to be required for efficient late gene expression. The induction of HSP70 by PML-II removal was specific for the HSPA1B gene among the HSP70 gene family and thus was not the consequence of a general stress response. Taken together, these data show that PML-II, through its various actions, has an overall negative effect on the Ad5 lifecycle.


Assuntos
Adenovírus Humanos/imunologia , Proteínas de Choque Térmico HSP70/metabolismo , Interferons/metabolismo , Proteína da Leucemia Promielocítica/metabolismo , Isoformas de Proteínas/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Imunidade Inata , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
5.
J Biol Chem ; 291(13): 6796-812, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26841862

RESUMO

The E3 transcription unit of human species C adenoviruses (Ads) encodes immunomodulatory proteins that mediate direct protection of infected cells. Recently, we described a novel immunomodulatory function for E3/49K, an E3 protein uniquely expressed by species D Ads. E3/49K of Ad19a/Ad64, a serotype that causes epidemic keratokonjunctivitis, is synthesized as a highly glycosylated type I transmembrane protein that is subsequently cleaved, resulting in secretion of its large ectodomain (sec49K). sec49K binds to CD45 on leukocytes, impairing activation and functions of natural killer cells and T cells. E3/49K is localized in the Golgi/trans-Golgi network (TGN), in the early endosomes, and on the plasma membrane, yet the cellular compartment where E3/49K is cleaved and the protease involved remained elusive. Here we show that TGN-localized E3/49K comprises both newly synthesized and recycled molecules. Full-length E3/49K was not detected in late endosomes/lysosomes, but the C-terminal fragment accumulated in this compartment at late times of infection. Inhibitor studies showed that cleavage occurs in a post-TGN compartment and that lysosomotropic agents enhance secretion. Interestingly, the cytoplasmic tail of E3/49K contains two potential sorting motifs, YXXΦ (where Φ represents a bulky hydrophobic amino acid) and LL, that are important for binding the clathrin adaptor proteins AP-1 and AP-2in vitro Surprisingly, mutating the LL motif, either alone or together with YXXΦ, did not prevent proteolytic processing but increased cell surface expression and secretion. Upon brefeldin A treatment, cell surface expression was rapidly lost, even for mutants lacking all known endocytosis motifs. Together with immunofluorescence data, we propose a model for intracellular E3/49K transport whereby cleavage takes place on the cell surface by matrix metalloproteases.


Assuntos
Adenoviridae/imunologia , Proteínas E3 de Adenovirus/química , Membrana Celular/imunologia , Células Epiteliais/imunologia , Fibroblastos/imunologia , Adenoviridae/química , Adenoviridae/patogenicidade , Proteínas E3 de Adenovirus/genética , Proteínas E3 de Adenovirus/imunologia , Motivos de Aminoácidos , Brefeldina A/farmacologia , Linhagem Celular Tumoral , Membrana Celular/virologia , Endossomos/imunologia , Endossomos/virologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Expressão Gênica , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunomodulação , Células Jurkat , Lisossomos/imunologia , Lisossomos/virologia , Dados de Sequência Molecular , Cultura Primária de Células , Estrutura Terciária de Proteína , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transdução de Sinais , Transfecção , Rede trans-Golgi/imunologia , Rede trans-Golgi/virologia
6.
mBio ; 7(1): e02184-15, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26814176

RESUMO

UNLABELLED: Modulation of host cell transcription, translation, and posttranslational modification processes is critical for the ability of many viruses to replicate efficiently within host cells. The human adenovirus (Ad) early region 4 open reading frame 3 (E4-ORF3) protein forms unique inclusions throughout the nuclei of infected cells and inhibits the antiviral Mre11-Rad50-Nbs1 DNA repair complex through relocalization. E4-ORF3 also induces SUMOylation of Mre11 and Nbs1. We recently identified additional cellular targets of E4-ORF3 and found that E4-ORF3 stimulates ubiquitin-like modification of 41 cellular proteins involved in a wide variety of processes. Among the proteins most abundantly modified in an E4-ORF3-dependent manner was the general transcription factor II-I (TFII-I). Analysis of Ad-infected cells revealed that E4-ORF3 induces TFII-I relocalization and SUMOylation early during infection. In the present study, we explored the relationship between E4-ORF3 and TFII-I. We found that Ad infection or ectopic E4-ORF3 expression leads to SUMOylation of TFII-I that precedes a rapid decline in TFII-I protein levels. We also show that E4-ORF3 is required for ubiquitination of TFII-I and subsequent proteasomal degradation. This is the first evidence that E4-ORF3 regulates ubiquitination. Interestingly, we found that E4-ORF3 modulation of TFII-I occurs in diverse cell types but only E4-ORF3 of Ad species C regulates TFII-I, providing critical insight into the mechanism by which E4-ORF3 targets TFII-I. Finally, we show that E4-ORF3 stimulates the activity of a TFII-I-repressed viral promoter during infection. Our results characterize a novel mechanism of TFII-I regulation by Ad and highlight how a viral protein can modulate a critical cellular transcription factor during infection. IMPORTANCE: Adenovirus has evolved a number of mechanisms to target host signaling pathways in order to optimize the cellular environment during infection. E4-ORF3 is a small viral protein made early during infection, and it is critical for inactivating host antiviral responses. In addition to its ability to capture and reorganize cellular proteins, E4-ORF3 also regulates posttranslational modifications of target proteins, but little is known about the functional consequences of these modifications. We recently identified TFII-I as a novel target of E4-ORF3 that is relocalized into dynamic E4-ORF3 nuclear structures and subjected to E4-ORF3-mediated SUMO modification. Here, we show that TFII-I is targeted by E4-ORF3 for ubiquitination and proteasomal degradation and that E4-ORF3 stimulates gene expression from a TFII-I-repressed viral promoter. Our findings suggest that the specific targeting of TFII-I by E4-ORF3 is a mechanism to inactivate its antiviral properties. These studies provide further insight into how E4-ORF3 functions to counteract host antiviral responses.


Assuntos
Adenovírus Humanos/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Elongação da Transcrição/metabolismo , Proteínas Virais/metabolismo , Células HeLa , Humanos , Proteólise , Sumoilação , Ubiquitina/metabolismo , Ubiquitinação
7.
Vaccine ; 33(48): 6641-9, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26529077

RESUMO

Pneumonia virus of mice (PVM) infection of BALB/c mice induces bronchiolitis leading to a fatal pneumonia in a dose-dependent manner, closely paralleling the development of severe disease during human respiratory syncytial virus infection in man, and is thus a recognised model in which to study the pathogenesis of pneumoviruses. This model system was used to investigate delivery of the internal structural proteins of PVM as a potential vaccination strategy to protect against pneumovirus disease. Replication-deficient recombinant human adenovirus serotype 5 (rAd5) vectors were constructed that expressed the M or N gene of PVM pathogenic strain J3666. Intranasal delivery of these rAd5 vectors gave protection against a lethal challenge dose of PVM in three different mouse strains, and protection lasted for at least 20 weeks post-immunisation. Whilst the PVM-specific antibody response in such animals was weak and inconsistent, rAd5N primed a strong PVM-specific CD8(+) T cell response and, to a lesser extent, a CD4(+) T cell response. These findings suggest that T-cell responses may be more important than serum IgG in the observed protection induced by rAd5N.


Assuntos
Adenovírus Humanos/genética , Portadores de Fármacos , Vírus da Pneumonia Murina/imunologia , Pneumonia Viral/veterinária , Infecções por Pneumovirus/prevenção & controle , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Vírus da Pneumonia Murina/genética , Pneumonia Viral/prevenção & controle , Análise de Sobrevida , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/imunologia , Vacinas Virais/genética
8.
J Virol ; 89(14): 7053-63, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25926634

RESUMO

UNLABELLED: The late phase of adenovirus gene expression is controlled by proteins made in the intermediate phase, including L4 proteins of 22,000- and 33,000-Da apparent molecular mass (L4-22K and -33K proteins) that are expressed initially from the L4 promoter (L4P). The L4P is activated by a combination of viral proteins and cellular p53 and is ultimately inhibited again by its own products. Here, we have examined the L4P of human adenovirus type 5 in detail and have defined its transcription start site, which our data suggest is positioned by a weak TATA box. Rather than contributing positively to promoter activity, a putative initiator element at the transcription start site acts as a target for negative regulation imposed on the L4P by cellular TFII-I. We show that this TFII-I inhibition is relieved by one of the previously defined viral activators of the L4P, the E4 Orf3 protein, which alters the pool of TFII-I in the cell. We also explore further the negative regulation of the L4P by its products and show that the L4-33K protein is more significant in this process than L4-22K. It is the combined actions of positive and negative factors that lead to the transient activation of the L4P at the onset of the late phase of adenovirus gene expression. IMPORTANCE: The adenovirus replication cycle proceeds through multiple phases of gene expression in which a key step is the activation of late-phase gene expression to produce proteins from which progeny particles can be formed. Working with human adenovirus type 5, we showed previously that two proteins expressed from the L4 region of the viral genome perform essential roles in moving the infection on into the late phase; these two proteins are produced by the action of a dedicated promoter, the L4P, and without them the infection does not proceed successfully to progeny generation. In this new work, we delineate further aspects of L4P activity and regulation. Understanding how the L4P works, and how it contributes to activation of the late phase of infection, is important to our understanding of natural infections by the virus, in which late gene expression can fail to occur, allowing the virus to persist.


Assuntos
Adenovírus Humanos/genética , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Regiões Promotoras Genéticas , Fatores de Transcrição TFII/metabolismo , Proteínas Virais/biossíntese , Linhagem Celular , Humanos , Sítio de Iniciação de Transcrição
9.
Mol Cell Biol ; 35(10): 1660-72, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25733689

RESUMO

To trigger type I interferon (IFN) responses, pattern recognition receptors activate signaling cascades that lead to transcription of IFN and IFN-stimulated genes (ISGs). The promyelocytic leukemia (PML) protein has been implicated in these responses, although its role has not been defined. Here, we show that PML isoform II (PML-II) is specifically required for efficient induction of IFN-ß transcription and of numerous ISGs, acting at the point of transcriptional complex assembly on target gene promoters. PML-II associated with specific transcription factors NF-κB and STAT1, as well as the coactivator CREB-binding protein (CBP), to facilitate transcriptional complex formation. The absence of PML-II substantially reduced binding of these factors and IFN regulatory factor 3 (IRF3) to IFN-ß or ISGs promoters and sharply reduced gene activation. The unique C-terminal domain of PML-II was essential for its activity, while the N-terminal RBCC motif common to all PML isoforms was dispensable. We propose a model in which PML-II contributes to the transcription of multiple genes via the association of its C-terminal domain with relevant transcription complexes, which promotes the stable assembly of these complexes at promoters/enhancers of target genes, and that in this way PML-II plays a significant role in the development of type I IFN responses.


Assuntos
Interferon beta/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/metabolismo , Proteínas Nucleares/química , Regiões Promotoras Genéticas , Proteína da Leucemia Promielocítica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/química , Proteínas Supressoras de Tumor/química
10.
J Virol ; 87(21): 11617-25, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23966406

RESUMO

During adenovirus infection, the emphasis of gene expression switches from early genes to late genes in a highly regulated manner. Two gene products, L4-22K and L4-33K, contribute to this switch by activating the major late transcription unit (MLTU) and regulating the splicing of its transcript. L4-22K and L4-33K expression is driven initially by a recently described L4 promoter (L4P) embedded within the MLTU that is activated by early and intermediate viral factors: E1A, E4 Orf3, and IVa2. Here we show that this promoter is also significantly activated by the cellular stress response regulator, p53. Exogenous expression of p53 activated L4P in reporter assays, while depletion of endogenous p53 inhibited the induction of L4P by viral activators. Chromatin immunoprecipitation studies showed that p53 associates with L4P and that during adenovirus type 5 (Ad5) infection, this association peaks at 12 h postinfection, coinciding with the phase of the infectious cycle when L4P is active, and is then lost as MLP activation commences. p53 activation of L4P is significant during Ad5 infection, since depletion of p53 prior to infection of either immortalized or normal cells led to severely reduced late gene expression. The association of p53 with L4P is transient due to the action of products of L4P activity (L4-22K/33K), which establish a negative feedback loop that ensures the transient activity of L4P at the start of the late phase and contributes to an efficient switch from early- to late-phase virus gene expression.


Assuntos
Adenovírus Humanos/genética , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Regiões Promotoras Genéticas , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular , Imunoprecipitação da Cromatina , Humanos , Ligação Proteica
11.
BMC Biotechnol ; 10: 92, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-21182761

RESUMO

BACKGROUND: Adenovirus serotype 5 (Ad5) has many favourable characteristics for development as a gene therapy vector. However, the utility of current Ad5 vectors is limited by transient transgene expression, toxicity and immunogenicity. The most promising form of vector is the high capacity type, which is deleted for all viral genes. However, these vectors can only be produced to relatively low titres and with the aid of helper virus. Therefore a continuing challenge is the generation of more effective Ad5 vectors that can still be grown to high titres. Our approach is to generate complementing cell lines to support the growth of Ad5 vectors with novel late gene deficiencies. RESULTS: We have used LoxP/Cre recombination mediated cassette exchange (RMCE) to generate cell lines expressing Ad5 proteins encoded by the L4 region of the genome, the products of which play a pivotal role in the expression of Ad5 structural proteins. A panel of LoxP parent 293 cell lines was generated, each containing a GFP expression cassette under the control of a tetracycline-regulated promoter inserted at a random genome location; the cassette also contained a LoxP site between the promoter and GFP sequence. Clones displayed a variety of patterns of regulation, stability and level of GFP expression. Clone A1 was identified as a suitable parent for creation of inducible cell lines because of the tight inducibility and stability of its GFP expression. Using LoxP-targeted, Cre recombinase-mediated insertion of an L4 cassette to displace GFP from the regulated promoter in this parent clone, cell line A1-L4 was generated. This cell line expressed L4 100K, 22K and 33K proteins at levels sufficient to complement L4-33K mutant and L4-deleted viruses. CONCLUSIONS: RMCE provides a method for rapid generation of Ad5 complementing cell lines from a pre-selected parental cell line, chosen for its desirable transgene expression characteristics. Parent cell lines can be selected for high or low gene expression, and for tight regulation, allowing viral protein expression to mirror that found during infection. Cell lines derived from a single parent will allow the growth of different vectors to be assessed without the complication of varying complementing protein expression.


Assuntos
Adenoviridae/genética , Linhagem Celular , Vetores Genéticos , Recombinação Genética , Técnicas de Cultura de Células , Teste de Complementação Genética , Humanos
12.
J Virol ; 84(14): 7096-104, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20444889

RESUMO

During human adenovirus 5 infection, a temporal cascade of gene expression leads ultimately to the production of large amounts of the proteins needed to construct progeny virions. However, the mechanism for the activation of the major late gene that encodes these viral structural proteins has not been well understood. We show here that two key positive regulators of the major late gene, L4-22K and L4-33K, previously thought to be expressed under the control of the major late promoter itself, initially are expressed from a novel promoter that is embedded within the major late gene and dedicated to their expression. This L4 promoter is required for late gene expression and is activated by a combination of viral protein activators produced during the infection, including E1A, E4 Orf3, and the intermediate-phase protein IVa2, and also by viral genome replication. This new understanding redraws the long-established view of how adenoviral gene expression patterns are controlled and offers new ways to manipulate that gene expression cascade for adenovirus vector applications.


Assuntos
Adenovírus Humanos/genética , Adenovírus Humanos/patogenicidade , Regulação Viral da Expressão Gênica , Regiões Promotoras Genéticas , Proteínas Virais/genética , Linhagem Celular , DNA Viral/genética , DNA Viral/metabolismo , Células HeLa , Humanos , Proteínas Virais/metabolismo , Replicação Viral/genética
13.
J Virol ; 83(7): 3049-58, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19176628

RESUMO

Adenoviruses express up to 20 distinct mRNAs from five major late transcription unit (MLTU) regions, L1 to L5, by differential splicing and polyadenylation of the primary transcript. MLTU expression is regulated at transcriptional and posttranscriptional levels. The L4-33K protein acts as a splicing factor to upregulate several MLTU splice acceptor sites as the late phase progresses. The L4 region also expresses a 22K protein whose sequence is related to the sequence of L4-33K. L4-22K is shown here also to have an important role in regulating the pattern of MLTU gene expression. An adenovirus genome containing a stop codon in the L4-22K open reading frame expressed low levels of both structural and nonstructural late proteins compared to the wild-type (wt) adenovirus genome; a decrease in intermediate proteins, IVa2 and IX, was also observed. However, early protein synthesis and replication were unaffected by the absence of L4-22K. Intermediate and late protein expression was restored to wt levels by L4-22K expressed in trans but not by L4-33K. Increased MLTU promoter activity, resulting from stabilization of the transcriptional activator IVa2 by L4-22K, made a small contribution to this restoration of late gene expression. However, the principal effect of L4-22K was on the processing of MLTU RNA into specific cytoplasmic mRNA. L4-22K selectively increased expression of penton mRNA and protein, whereas splicing to create penton mRNA is known not to be increased by L4-33K. These results indicate that L4-22K plays a key role in the early-late switch in MLTU expression, additional to and distinct from the role of L4-33K.


Assuntos
Adenoviridae/fisiologia , Regulação Viral da Expressão Gênica , Proteínas Virais/metabolismo , Replicação Viral , Códon sem Sentido , Teste de Complementação Genética
14.
J Gen Virol ; 90(Pt 1): 95-104, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19088278

RESUMO

Human adenovirus type 5 infection causes the disruption of structures in the cell nucleus termed promyelocytic leukaemia (PML) protein nuclear domains or ND10, which contain the PML protein as a critical component. This disruption is achieved through the action of the viral E4 Orf3 protein, which forms track-like nuclear structures that associate with the PML protein. This association is mediated by a direct interaction of Orf3 with a specific PML isoform, PMLII. We show here that the Orf3 interaction properties of PMLII are conferred by a 40 aa residue segment of the unique C-terminal domain of the protein. This segment was sufficient to confer interaction on a heterologous protein. The analysis was informed by prior application of a bioinformatic tool for the prediction of potential protein interaction sites within unstructured protein sequences (predictors of naturally disordered region analysis; PONDR). This tool predicted three potential molecular recognition elements (MoRE) within the C-terminal domain of PMLII, one of which was found to form the core of the Orf3 interaction site, thus demonstrating the utility of this approach. The sequence of the mapped Orf3-binding site on PML protein was found to be relatively poorly conserved across other species; however, the overall organization of MoREs within unstructured sequence was retained, suggesting the potential for conservation of functional interactions.


Assuntos
Adenoviridae/fisiologia , Proteínas E4 de Adenovirus/metabolismo , Proteínas Nucleares/metabolismo , Mapeamento de Interação de Proteínas , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Biologia Computacional/métodos , Sequência Conservada , Humanos , Dados de Sequência Molecular , Proteína da Leucemia Promielocítica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/genética , Alinhamento de Sequência
15.
J Virol ; 80(6): 3042-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501113

RESUMO

Nuclear domain 10 (ND10s), or promyelocytic leukemia protein (PML) nuclear bodies, are spherical nuclear structures that require PML proteins for their formation. Many viruses target these structures during infection. The E4 Orf3 protein of adenovirus 5 (Ad5) rearranges ND10s, causing PML to colocalize with Orf3 in nuclear tracks or fibers. There are six different PML isoforms (I to VI) present at ND10s, all sharing a common N terminus but with structural differences at their C termini. In this study, PML II was the only one of these six isoforms that was found to interact directly and specifically with Ad5 E4 Orf3 in vitro and in vivo; these results define a new Orf3 activity. Three of a series of 18 mutant Orf3 proteins were unable to interact with PML II; these were also unable to cause ND10 rearrangement. Moreover, in PML-null cells that contained neoformed ND10s comprising a single PML isoform, only ND10s formed of PML II were rearranged by Orf3. These data show that the interaction between Orf3 and PML II is necessary for ND10 rearrangement to occur. Finally, Orf3 was shown to self-associate in vitro. This activity was absent in mutant Orf3 proteins that were unable to form tracks and to bind PML II. Thus, Orf3 oligomerization may mediate the formation of nuclear tracks in vivo and may also be important for PML II binding.


Assuntos
Proteínas E4 de Adenovirus/metabolismo , Adenovírus Humanos/patogenicidade , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas E4 de Adenovirus/química , Proteínas E4 de Adenovirus/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Estruturas do Núcleo Celular/patologia , Imunofluorescência , Humanos , Imunoprecipitação , Camundongos , Dados de Sequência Molecular , Proteína da Leucemia Promielocítica
16.
Exp Cell Res ; 307(1): 109-17, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15922731

RESUMO

The PML protein is a defining constituent of subnuclear structures known as ND10. PML is expressed as a series of primary sequence isoforms through alternative RNA processing. Expression of each of six distinct PML isoforms that differed in their C-terminal domains caused reproducible differences in the number, size, and shape of ND10 in both transformed cell lines and diploid fibroblasts. In each case, PML from the endogenous genes was reorganized to participate with the exogenously expressed PML in the new configuration of ND10. Variation in ND10 number is known to occur during the cell cycle; however, the cell cycle distribution of the transfected cells that displayed these altered ND10 was similar for all six PML isoforms. Given our findings, the precise level of expression of the different PML isoforms under particular physiological conditions will be an important determinant of ND10 organization and function and is a potential point of regulation of PML/ND10 function.


Assuntos
Estruturas do Núcleo Celular/fisiologia , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Estruturas do Núcleo Celular/metabolismo , Citometria de Fluxo , Corantes Fluorescentes , Humanos , Indóis , Leucemia Promielocítica Aguda/patologia , Microscopia Confocal , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Plasmídeos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Transfecção
17.
Oncogene ; 23(27): 4662-72, 2004 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15107834

RESUMO

Mucosal human papillomaviruses (HPVs) are the causative agents of a number of human pathologies, including benign condylomas, as well as of the majority of cervical cancers and their high-grade precursor lesions. Although the viral E6 protein is known to be essential for driving malignant progression of HPV-infected cells, there are still many uncertainties about its mode of action. In this study, we have analysed the intracellular distribution of the E6 oncoproteins from the high-risk HPV-18 and the low-risk HPV-11. We show that both E6 proteins localize within the nucleus in nuclear bodies that are confocal with the promyelocytic leukaemia (PML) protein. Using a panel of different PML isoforms, we demonstrate specific co-localization between the E6 proteins and PML isoforms I-IV, but not with PML isoforms V and VI. We also demonstrate the interaction between E6 and a subset of PML isoforms in vivo. As a consequence of this interaction, the insoluble form of PML IV is destabilized by HPV-18 E6 through a proteasome-dependent pathway. Interestingly, both HPV-11 E6 and HPV-18 E6 can readily overcome PML IV-induced cellular senescence in primary cells. These results show separable functions for different PML isoforms that are specifically targeted by the HPV E6 oncoproteins.


Assuntos
Núcleo Celular/metabolismo , Fibroblastos/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/metabolismo , Isoformas de Proteínas/metabolismo , Processamento Alternativo , Animais , Anticorpos Monoclonais/metabolismo , Western Blotting , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Cisteína Endopeptidases/metabolismo , Fibroblastos/citologia , Fibroblastos/virologia , Técnica Direta de Fluorescência para Anticorpo , Humanos , Microscopia Confocal , Complexos Multienzimáticos/metabolismo , Proteínas Oncogênicas Virais/análise , Proteínas Oncogênicas Virais/genética , Testes de Precipitina , Complexo de Endopeptidases do Proteassoma , Isoformas de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos
18.
J Virol ; 78(7): 3470-9, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15016870

RESUMO

The efficiency and specificity of gene transfer with human adenovirus (hAd)-derived gene transfer vectors would be improved if the native viral tropism could be modified. Here, we demonstrate that the minor capsid protein IX (pIX), which is present in 240 copies in the Ad capsid, can be exploited as an anchor for heterologous polypeptides. Protein IX-deleted hAd5 vectors were propagated in hAd5 helper cells expressing pIX variants, with heterologous carboxyl-terminal extensions of up to 113 amino acids in length. The extensions evaluated consist of alpha-helical spacers up to 75 A in length and to which peptide ligands were fused. The pIX variants were efficiently incorporated into the capsids of Ad particles. On intact particles, the MYC-tagged-pIX molecules were readily accessible to anti-MYC antibodies, as demonstrated by electron microscopic analyses of immunogold-labeled virus particles. The labeling efficiency improved with increasing spacer length, suggesting that the spacers lift and expose the ligand at the capsid surface. Furthermore, we found that the addition of an integrin-binding RGD motif to the pIX markedly stimulated the transduction of coxsackievirus group B and hAd receptor-deficient endothelioma cells, demonstrating the utility of pIX modification in gene transfer. Our data demonstrate that the minor capsid protein IX can be used as an anchor for the addition of polypeptide ligands to Ad particles.


Assuntos
Adenoviridae/fisiologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Engenharia Genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Adenoviridae/genética , Animais , Western Blotting , Proteínas do Capsídeo/genética , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Ligantes , Camundongos , Microscopia Imunoeletrônica , Mutação/genética , Ligação Proteica , Transporte Proteico , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/genética , Especificidade por Substrato
19.
J Virol ; 78(4): 1782-91, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14747543

RESUMO

The adenovirus major late transcription unit (MLTU) encodes multiple proteins from five regions, L1 to L5, through differential splicing and polyadenylation. MLTU expression is temporally regulated; only a single product from L1 (52/55K) is expressed prior to replication, but a subsequent switch, the mechanism of which has not been defined, leads to full expression that encompasses L1 IIIa and all L2 to L5 products. Transfection of a plasmid containing the complete MLTU gave a full array of proteins in proportions similar to those in a late infection, and in a time course, the temporal pattern of expression in a natural infection was reproduced. However, a plasmid truncated after the L3 poly(A) site exclusively expressed the L1 52/55K protein and was defective in the switch to full gene expression from L1 to L3. The L4 33K protein, supplied in trans, was sufficient to upregulate cytoplasmic mRNA for MLTU products characteristic of the late pattern of expression to levels comparable to those produced by the full-length MLTU. There was a corresponding increase in expression of the L1 IIIa, L2, and L3 proteins, except hexon. Hexon protein expression additionally required both the L4 100K protein in trans and sequences downstream of the L3 poly(A) site in cis. These results indicate that induction of L4 protein expression is a key event in the early-late switch in MLTU expression, which we propose is precipitated by small amounts of L4 expression in a feed-forward activation mechanism.


Assuntos
Adenovírus Humanos/metabolismo , Elementos Facilitadores Genéticos , Regulação Viral da Expressão Gênica , Transcrição Gênica , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Adenovírus Humanos/genética , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Humanos , Plasmídeos , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , RNA Viral , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética
20.
J Gen Virol ; 84(Pt 2): 259-268, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12560556

RESUMO

Human adenovirus serotype 5 encodes three proteins, E1b 55K, E4 Orf3 and E4 Orf6, which interact with each other and with components of the nucleus to regulate mRNA processing and export, viral DNA replication and p53-dependent apoptosis. Previous studies have shown that, during wild-type infection, 55K associates initially with structures termed ND10, which are sites of localization of the promyelocytic leukaemia protein, and then moves, dependent upon its interaction with Orf6, to the establishing virus replication centres. Absence of either Orf3 or Orf6 affects the localization of 55K and so may affect its function. In this study, the influence of Orf3 and Orf6 expression on the association of 55K with the insoluble matrix fraction of the nucleus and with ND10 particularly was examined. Overexpression of Orf6 was sufficient to block the association of 55K with this fraction, irrespective of the presence of Orf3. This effect depended upon the two proteins being able to interact. However, the association of 55K with ND10, which persists throughout infection in the absence of Orf6, required Orf3 to be present, thus distinguishing two subsets of matrix-associated 55K. A modified form of 55K, formation of which was blocked by mutating the known site of SUMO-1 attachment, was more abundant in the absence of Orf6 but unaffected by the absence of Orf3. Thus, this modification is favoured when 55K remains associated with the matrix but does not correlate with its stable association with ND10, many components of which are modified by SUMO-1.


Assuntos
Proteínas E1B de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Adenovírus Humanos/patogenicidade , Regulação Viral da Expressão Gênica , Matriz Nuclear/metabolismo , Proteína SUMO-1/metabolismo , Linhagem Celular , Células HeLa , Humanos , Fases de Leitura Aberta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...